Abstract

The prognosis for patients with brain metastasis remains dismal despite intensive therapy including surgical resection, radiotherapy, chemo-, targeted, and immunotherapy. Thus, there is a high medical need for new therapeutic options. Recent advances employing high-throughput and spatially resolved single-cell analyses have provided unprecedented insights into the composition and phenotypes of the diverse immune cells in the metastatic brain, revealing a unique immune landscape starkly different from that of primary brain tumors or other metastatic sites. This review summarizes the current evidence on the composition and phenotypes of the most prominent immune cells in the brain metastatic niche, along with their dynamic interactions with metastatic tumor cells and each other. As the most abundant immune cell types in this niche, we explore in detail the phenotypic heterogeneity and functional plasticity of tumor-associated macrophages, including both resident microglia and monocyte-derived macrophages, as well as the T-cell compartment. We also review preclinical and clinical trials evaluating the therapeutic potential of targeting the immune microenvironment in brain metastasis. Given the substantial evidence highlighting a significant role of the immune microenvironmental niche in brain metastasis pathogenesis, a comprehensive understanding of the key molecular and cellular factors within this niche holds great promise for developing novel therapeutic approaches as well as innovative combinatory treatment strategies for brain metastasis.

Brain metastases (BrM) arise in around 25% of all adult cancer patients, constituting the most common brain neoplasm in adults with the highest prevalence in nonsmall cell lung cancer (NSCLC), breast cancer, and melanoma.1 Despite intensive therapy including surgical resection,2 radiotherapy,3 and drug treatment,4 patient survival rarely exceeds 2 years.5 Beyond cancer cell-intrinsic mechanisms, the tumor microenvironment (TME) comprising stromal and immune cells has become the focus of intensive investigation in systemic, as well as central nervous system (CNS) tumors.6 The significance of the brain metastatic microenvironment (brain-MME) has been demonstrated in several studies, showing that successful brain colonization by disseminated cancer cells depends on tumor cell–immune–stroma interactions.6 The heterogeneous, immunosuppressive nature of the brain-MME has been linked to therapeutic failure,7 and immune microenvironment signatures correlate with the prognosis of patients with BrM.8

The brain consists of a unique cellular landscape exhibiting remarkable differences in comparison to other tissues frequently associated with metastasis. Beyond a diversity of interconnected neurons, the brain architecture is characterized by the presence of glia cells such as astrocytes and oligodendrocytes, pericytes, ependymal cells, and endothelial cells, as well as a unique extracellular matrix.6 The role of these stromal components of the brain-MME, including astrocytes, neurons, and the blood–brain barrier (BBB), has been recently reviewed elsewhere.9

The brain-MME is defined by a rich diversity of immune cells. Historically assumed to be immune privileged due to the isolation from peripheral immune cells by the BBB, research over the past years has challenged this narrative by revealing neuroimmune interactions in health and disease. This is particularly true in the context of brain tumors that are accompanied by a compromised BBB allowing for a substantial influx of peripheral immune cells. Consequently, the immune brain-MME is not only characterized by resident microglia, macrophages, and dendritic cells (DCs) but also by infiltrating monocytes, blood-derived macrophages, neutrophils, and lymphocytes (Figure 1).

Simplified overview of tumor–immune interactions in the brain metastatic microenvironment. This figure illustrates the well-established (consensus) interactions between various immune cell subsets and metastatic tumor cells, providing an overview of the immune dynamics in brain metastasis. Arrows indicate supportive interactions, inhibitory arcs indicate inhibition, and dotted lines suggest interactions with only limited evidence. Myeloid and lymphatic immune cell populations reveal dynamic interactions with tumor cells in the metastatic microenvironment, impacting treatment response, recurrence, and patient survival. Beyond, cellular intrinsic factors such as tumor genotype and immune cell ontogeny, as well as extrinsic ones such as patient demographics and pretreatment, shape the observed immune cell phenotypes.
Figure 1.

Simplified overview of tumor–immune interactions in the brain metastatic microenvironment. This figure illustrates the well-established (consensus) interactions between various immune cell subsets and metastatic tumor cells, providing an overview of the immune dynamics in brain metastasis. Arrows indicate supportive interactions, inhibitory arcs indicate inhibition, and dotted lines suggest interactions with only limited evidence. Myeloid and lymphatic immune cell populations reveal dynamic interactions with tumor cells in the metastatic microenvironment, impacting treatment response, recurrence, and patient survival. Beyond, cellular intrinsic factors such as tumor genotype and immune cell ontogeny, as well as extrinsic ones such as patient demographics and pretreatment, shape the observed immune cell phenotypes.

Myeloid Cells

As an integral part of both innate and adaptive systemic immunity, myeloid cells including macrophages, neutrophils and DCs are increasingly recognized as a key factor in brain physiology and as central regulators of metastasis.

Tumor-Associated Macrophages

Tumor-associated macrophages (TAMs) constitute one of the most abundant cell types found in the brain-MME, accounting for around 30%–40% of nontumor cells.10–12 TAMs comprise 2 subpopulations defined by tissue origin: CNS-resident TAMs and infiltrating, monocyte-derived macrophages (MDMs).6

CNS-resident TAMs can be subdivided into 2 populations: the predominant tissue-resident macrophages microglia, and a group of macrophages populating the CNS borders (choroid plexus, perivascular spaces, meninges), collectively termed border-associated macrophages (BAMs). Microglia originate from yolk-sac–derived progenitors during embryological development and are maintained by self-renewal rather than peripheral monocyte influx. Functioning as the brain’s primary immune cells, microglia are paramount for local immune defence owing to a diverse set of receptors including chemokine, pattern recognition, and purinergic receptors, termed the microglia sensome. Microglia possess substantial phenotypic plasticity, as their transcriptional differences have been described in relation to regional differences in the brain.13 Also stemming from yolk sac as well as fetal liver origins,14 BAMs serve as the tissue-resident macrophagic population at the CNS border regions. BAMs have been described to contain increased antigen presentation capacity compared with microglia, potentially due to localizing at brain–periphery interfaces.14 Notwithstanding, little is known about the selective contribution of BAMs in BrM,7 and thus far, microglia are considered the primary source of CNS-resident TAMs.

Monocytes, not physiologically detected in the brain besides small populations residing in the meninges, are recruited to the brain through chemokine release during BrM9 and differentiate into MDMs intratumorally.6

Heterogeneity of TAMs

.—Understanding the functional contributions of TAM subpopulations by ontogeny has been challenged with difficulty in distinguishing TAM subsets.9,15 Classical markers used to differentiate microglia from MDMs such as CX3CR1, CD45, P2RY12, and TMEM119 fail to reliably delineate the two populations, particularly after immune-shaping activities of metastatic cells.16,17

Extending the identification of subsets beyond the use of lineage tracing and conditional genomic modifications in mouse models, several markers have been proposed for better cell separation over the last years. For example, MDMs have been suggested to be identifiable by the expression of CD49D (ITGA4),16 CD141, and ICAM.10 Discrepancy concerning the regional distribution of TAM subsets in BrM likely reflects the longstanding difficulty to finely disentangle resident microglia from infiltrating MDMs. Employing ITGA4 as an MDM marker revealed that microglia-derived TAMs predominantly populate the tumor margin while being largely absent from the tumor core. MDMs in turn were detected primarily in the tumor core, and special subsets of MDMs expressing markers of antigen presentation are localized in the proximity of tumor blood vessels.10

Additionally, TAM composition is highly dynamic during the course of disease. In early, small BrM lesions, the TAM compartment mainly consists of microglia, continued influx of monocytes over disease progression shifting the composition towards a predominance of MDMs.18,19 Notably, the described monocytic influx occurs independently of therapeutic disruptions of the BBB and rather constitutes a BrM inherent feature.16

Phenotypic plasticity of TAMs in BrM

.—In line with the phenotypic variance of microglia and macrophages in health and disease, TAMs in BrM exhibit several notable phenotypic characteristics. In general, education by the brain-MME induces stark phenotypic convergence of TAMs independent of ontogeny.17 Although there is no clear pattern of a traditionally defined M1 or M2 phenotype,11 the pro- and anti-inflammatory signatures of TAMs in BrM have been comprehensively described and correlated with clinical outcome. For example, an inflammation-associated IFNγ signature is correlated with prolonged survival in lung cancer BrM patients.20

BrM-associated microglia show an activated stellate and reactive amebic morphology throughout the course of disease.10,18,21 Beyond an observed downregulation of the homeostatic microglial markers CX3CR1, P2RY12, and TMEM119, microglia in the brain-MME show little similarity to other neurological disease-associated microglia.17 Although microglial phenotype generally does not align well with a reductionist M1-M2 or anti-/pro-inflammatory dichotomy,9 BrM-associated microglia population display an immature and inflammatory phenotype,22 with an induced expression of pro-inflammatory cytokines, interleukins, and TNF, as well as the complement cascade and S100 family proteins.11,17,18,23 Besides, enrichment of exosomal genes, TYRO3/AXL/MER receptor signaling, and lipid metabolism have also been reported.18,23 Increased gene expression of innate immune sensing genes such as Toll-like receptor signaling and activation markers (CD14, CD163, and CD64) has been described.10 Notably, upregulated expression of several anti-inflammatory genes has also been associated with microglia in BrM, including LGALS1/3 and IFITM3,17 VEGFA/B, IGF1, HGF, and FGF20.24

BrM-associated MDMs constitute a more transcriptionally heterogeneous group inclusive of subsets such as microglia-similar, phagocytic/lipid-, hypoxic-glycolytic macrophages, or those expressing anti-inflammatory or IFN signatures.7 This diversity of MDM subsets could be reflected in functional differences regarding immunological capacity and tumor support.7 Interactions of MDMs with tumor cells and the brain-MME can induce downregulation of CCR2, FCN1, and VCAN while upregulating macrophage markers such as CD163, MERTK, and C1QA during the differentiation of monocytes into macrophages.10,17 In contrast to microglia, BrM-MDMs display more induced markers of the alternative activation (M2-like) phenotype including CCL17, CCL22, CD163, and TLR1.18 Several anti-inflammatory molecules, such as FCGR2B, CLEC10A, ARG1, CD39, TGFβ, IL10, IL1RN, CD155, MSR1, and MHC-II, are abundantly expressed in MDMs.9,11,16 Consequently, BrM-MDMs are frequently associated with antigen processing.16,17,25 Phenotypic markers indicating an increased phagocytic and antigen cross-presentation ability similar to DCs such as CD1C, CD1B, and CD207 have been identified in MDM population.11 Notwithstanding, the phenotype of MDMs remains highly plastic with the increased expression of several pro-inflammatory markers including TSPO, ISG15, IFITM2, ANXA2, IRF7, S100A11, LGALS, and IL1b,17 and the cytokines CCL8, CCL13, CCL17, and CCL18.11

Alternative clustering has identified an APOE-expressing TAM subset that shows the strong enrichment of antigen processing and presentation, which also expresses immunomodulatory molecules such as the complement C1Q chains, SPP1, and HLA-related molecules. An S100A8 expressing subset characterized by the high expression of S100A family genes CXCL8 and FCN1, and the low expression of HLA-related genes has also been reported, which resemble myeloid-derived suppressor cells with the enrichment of cytokine-mediated signaling and response to IL1.26

Functional contribution of TAMs along the metastasis cascade

.—TAMs have been implicated as integral players in the pathogenesis of BrM (Figure 2). Already early during metastatic dissemination, endothelial-derived DKK1 release triggered by lung cancer-derived exosomes was shown to induce a protumorigenic phenotype in microglia, contributing to the establishment of the premetastatic niche.27 In agreement, breast cancer cell-derived ANXA1 steered microglial migration prior to metastatic establishment and ANXA1 induced microglial activation via STAT3.28 Aiding early metastatic dissemination to the brain, microglia activation by IL6 and CSF1 facilitates tumor cell BBB transmigration.25,29 In experimental melanoma models, intravital imaging established an involvement of activated TAMs in metastasis formation by disrupting BBB integrity via MMP3.30 Depletion of TAMs with an immunosuppressive phenotype by mannosylated clodronate liposomes in animal models further abrogated breast cancer BrM burden during early disease stages.31

Functional contribution of TAMs along the metastasis cascade. Microglia contribute to the establishment of the premetastatic niche and aid tumor cell extravasation by altering the BBB. During early metastatic establishment, microglia are reprogrammed by tumor-derived signals to facilitate invasion and colonization. Monocytes attracted to the metastatic site differentiate into MDMs further aiding metastatic growth. Finally, in the surrounding of established metastases, TAMs attract lymphocytes and convey immunosuppression.
Figure 2.

Functional contribution of TAMs along the metastasis cascade. Microglia contribute to the establishment of the premetastatic niche and aid tumor cell extravasation by altering the BBB. During early metastatic establishment, microglia are reprogrammed by tumor-derived signals to facilitate invasion and colonization. Monocytes attracted to the metastatic site differentiate into MDMs further aiding metastatic growth. Finally, in the surrounding of established metastases, TAMs attract lymphocytes and convey immunosuppression.

TAMs play an established role in brain invasion, particularly by virtue of their interactions with tumor cells during the invasion/metastasis cascade. Upon extravasation of singular tumor cells, microglia get recruited, resulting in direct contact and communication with the cancer cells.21,25 Microglia have been shown to aid brain invasion in a WNT pathway-dependent manner, which was reversible by either WNT inhibition or LPS-elicited activation.32 Furthermore, tumor cell invasion-inflicted perturbations in the brain structure, which are associated with microglial damage response, can spark enhanced tumor invasion in a CXCR4-dependent manner.15 This microenvironmental determination of tumor cell invasion is in line with an overall lack of association between primary tumor type and invasion patterns.33

Finally, several mechanisms facilitating colonization also involve TAMs. Microglia expressing CD74 were shown to promote metastatic growth via midkine (MDK) secretion.34 PI3K was additionally identified as a central regulator of TAM-mediated colonization, PI3K inhibition altering metastatic growth.35 Moreover, cathepsin S released by MDMs was implicated in enhancing brain colonization.36 Colonization was further assisted by tumor-derived IL6, which abrogated microglial inflammation via the JAK2/STAT3 axis in NSCLC BrM. Elevated serum levels of IL6 correlate with BrM occurrence and survival.29 A trilateral interaction has been characterized, in which astrocytic exosomes inhibit BrM PTEN expression, thereby inducing alterations in chemokine secretion by tumor cells, which in turn enhance the attraction of bone marrow-derived macrophages to the metastatic site.37 In addition, increased myeloid cell content has been correlated with immunosuppression, increased metastatic cell proliferation, as well as therapeutic failure.38 Similarly, the intratumoral density of CD163+ TAMs correlates with shorter survival.39 In line, microglia-conditioned media exhibit tumor-supportive properties in vitro.40

As part of the innate immune system with intrinsic antitumor capacity, phagocytosis of early extravasated tumor cells has been observed in TAMs.19 Notably, the phagocytic capacity of TAMs was lost during disease progression, accompanied by TAM activation and MDM influx. Comparable phagocytic and tumorolytic capacity have been described using intravital imaging.41 LPS-induced activation of both microglia and MDMs could enable tumor cell killing in vivo and in vitro, underlining the overall potential for an effective antitumor immunity of TAMs.40 Interestingly, tamoxifen treatment in an animal model of breast cancer ameliorated BrM by shifting microglia toward a more phagocytic phenotype.42

Beyond the well-described involvement of TAMs in BrM establishment, their functional importance in orchestrating the niche immunity via multifaceted interactions with other immune cells, encompassing not only pro-tumor but also antitumor functions, has been a controversial field of debate. Pointing toward antitumor immunity orchestrated by microglial TAMs in concert with other immune cells, animals lacking microglia presented with enhanced tumor progression, paralleled by decreased natural killer (NK) cell and T-cell responses.23 In line, microglial markers correlate with increased survival in breast cancer BrM.23 Furthermore, TAM-secreted chemokines such as CCL3/4 are central for lymphocyte recruitment.24 Interestingly, the elevated abundance of microglia and monocytes correlated with reduced local recurrence and leptomeningeal involvement in melanoma BrM.12 This finding indicates not only a role for microglia in antitumor immunity but also a potential protective effect of monocyte infiltration, supporting the notion that naive monocytes still retain immunocompetence, unlike the tumor-shaped MDMs.7

Conversely, evidence points toward TAM involvement in shaping the immunosuppressive nature of the brain-MME via secretion of factors such as galectin-1,24 Cystatin C,43 or the production of immunosuppressive metabolites via IDO upregulation.44 These changes are believed to limit antitumor immunity by altering T-cell–mediated immunity: increasing CD8+ T-cell exclusion, reducing T-cell activation, and increasing immune checkpoint expression24 while facilitating regulatory T-cell (T-reg) attraction.45 Microglial CX3CR1 expression loss, as typically observed in BrM, can induce a microglial support to BrM outgrowth by recruiting CD68+ myeloid cells with high expression of checkpoint markers PD-L1, VISTA, which hamper T-cell–mediated immunity.17 This aligns with transcriptomic studies implicating TAMs, particularly MDMs, in immunosuppression and altered T-cell function.16,46 Conversely, metastatic-derived factors such as neurotrophin-347 and amyloid-β48 have been demonstrated to foster immunosuppression by limiting microglial activation and phagocytic capacity.

Altogether, the evidence presented suggests a multifaceted involvement of TAMs in pro- and antitumor immunity. While limited evidence points towards an effective antitumor immune response by resident microglia during the initial steps of BrM, the educational influence of tumor-intrinsic and environmental factors potentially hijacks microglial-mediated immune signaling to facilitate metastasis outgrowth. While available evidence indicates a more tumorigenic function of infiltrating MDMs, insufficient data exist to clearly delineate the immunological contribution.

Neutrophils

Neutrophils represent 50%–70% of the myeloid-derived circulating leukocytes in human blood and are involved in innate immune response against invading pathogens. They also represent most of the inflammatory cells in solid tumors with a high intratumor density and are increasingly identified as key drivers of tumor progression by dictating TME composition and tumor vascularization.49 Neutrophil increase can be observed in an elevated neutrophil-to-lymphocyte ratio in peripheral blood,50 which has been associated with poorer survival postsurgical resection.51 Phenotypically, BrM-associated neutrophils show an induction of immune and inflammatory pathways including TNFα signaling, whereas ROS production is reduced.50 BrM-associated neutrophils are characterized by high expression of infiltration and transmigration markers such as ITGA3, CD177, CD15, and CD11b.11,50 In line with this immune-suppressive phenotype, neutrophils exhibit high expression of PD-L1, which was most strongly detected in vicinity to PD1+ CD8+ T cells, implicating a role for neutrophils in hampering antitumor immunity.50 Immunosuppressive ARG1- and PD-L1-expressing neutrophils were shown to drive metastatic progression following attraction by tumor-derived G-CSF.52 Beyond immune-related functions, neutrophils have been implicated in angiogenesis, particularly localizing close to blood vessels within the tumor mass.50,53 During early steps of BrM, the primary tumor induces astrocyte-mediated inflammation, which in turn causes an attraction of neutrophils to the metastatic niche.54 Furthermore, granulocytes have been implicated in metastatic seeding following their attraction to the premetastatic niche by elevated levels of S100A8/9.55 BrM-associated neutrophils also show elevated expression of S100A9,50 potentially instigating a self-reinforcing cycle of neutrophil attraction. Intratumoral neutrophils express markers of recruitment of anti-inflammatory neutrophils (CXCL17, ADORA2A, and MET).11,50 Based on these observations, strategies aiming at targeting tumor neutrophil infiltration by pharmacologically blocking chemokine signaling (eg, blocking IL8 and CXCL1)49 or neutrophil-derived substances are currently viewed as valuable therapeutic options.

Dendritic cells

Acting as professional antigen-presenting cells, DCs are pivotal players initiating and regulating T-cell–elicited antitumor immune responses. A cluster of CD163 and CD14 expressing DCs was identified, which exhibits elevated chemokine expression, suggesting a role for T-cell recruitment.56 Similarly, a cluster of type 3 DCs involved in T-cell activation and MHC expression was revealed in melanoma BrM.57 Further exploration of patient samples of BrM of diverse tumor origin revealed a prevalence of type 2 DCs that subclustered into 2 fractions with mutually exclusive expression levels of either migratory receptors or antigen presentation.26

Lymphoid Compartment

The brain contains a limited number of lymphocytic cells under physiological conditions, except for circumscribed functions of patrolling lymphocytes in the CNS border such as meninges and choroid plexus.58 In stark contrast, lymphocytes represent the most prevalent immune cell type in the brain-MME, constituting 35%–65% of all cells.10,11 The predictive value of tumor-associated lymphocyte abundance for patient prognosis is still disputed; however, the elevated presence of some lymphocyte subsets, particularly CD8+ T cells and cytotoxic lymphocytes, is associated with patient prognosis.59 Conversely, progression under treatment was associated with lower lymphocyte density.60

T Cells

T-cells constitute the majority of BrM-associated lymphocytes,10,11 containing the largest phenotypic diversity of all immune cells in the brain-MME.26 Blood vessels in the vicinity of metastatic cells express endothelial adhesion molecules such as VCAM1 and ICAM1 in different BrM entities,61 which aligns with reports of increasing abundance of both CD8+ and CD4+ T-cell subsets with increasing tumor grade.10

Infiltrating T-cells exhibit a mixed composition, consisting of both CD8 and CD4-expressing populations. Studies report a predominance of CD4+ T cells, with a high abundance of T-regs,10,62 spanning a phenotypic spectrum from effector to central memory T cells.26 Some reports describe the high accumulation of naïve and memory T cells, including both CD8+ effector memory and central memory T cells.10,20,26 Notably, effector memory T-cells constitute the majority of CD8+ T cells, revealing a cytotoxic phenotype.20,26 When tracking T-cell receptor (TCR) diversity, TCR clonal expansion occurred in cytotoxic T-cell fractions showing potential tumor reactivity, and singular TCR clones predominantly found in naïve T cells and T-regs.20 The T-cell compartment in BrM is characterized by an activated and exhausted phenotype.10 Beyond extensive PD1 expression, other checkpoint molecules such as TIM3, CTLA4, TIGIT, and LAG3 are abundantly expressed.7 Interestingly, when investigating the organizational dimension underlying T-cell diversity, the activation state was revealed to orchestrate the phenotypic heterogeneity.26 Similar to phenotypic changes observed in TAMs, changes in the activation state of T-cell subsets were paralleled by metabolic profiles.26

Throughout BrM, dynamic interactions of inflammatory TAMs and T-cells have been reported. Preclinical evidence points towards an adaptation of an immunosuppressive phenotype of T cells upon contact with microglia.7 In contrast, T cells have recently been suggested to be responsible for inducing antigen presentation in tumor-associated microglia that are else restricted to interferon response.23 Interestingly, TAMs isolated from patients identified to possess T-cell clones with potential antitumor capacity were shown to highly express both these two programs alongside strong T-cell recruitment capacity.63

CD8 + T-cells

.—In line with the overall immunosuppressive nature of the T-cell compartment in BrM, CD8+ T-cells exhibit an exhausted signature, expressing PD1 and other co-inhibitory molecules such as CTLA4 and TIM3,25,60 with only little overlap with peripheral CD8+ T-cells. Beyond the PD1+ cluster, a memory-like CD8+ subcluster was revealed.60 These CD8+ subclusters also show spatially defined distribution based on antigen specificity, with exhausted CD8+ T-cells being most prevalent in the tumor core, whereas CD8+ memory T-cells were mainly present in the stroma-rich tumor margins.60 Other studies have clustered infiltrating CD8+ T-cells phenotypically into a conventional cytotoxic and an interferon (IFN) signaling expressing cluster.7 Notably, a significant proportion of tumor-infiltrating CD8+ T-cells was characterized as reactive to nontumor, microbial antigens rather than tumor-specific ones.60 In support, Wischnewski et al. reported that T-cells with low tumor-reactivity showed larger overlap with peripheral populations than tumor-antigen–specific CD8+ T-cells. Furthermore, a population of TOX-expressing CD8+ T-cells has recently been identified in melanoma BrM, which exhibits lower immune checkpoint marker expression compared with matched primary tumor samples.64 Finally, several CD8+ subsets not only express markers of inhibition and activation but also co-express stimulatory receptors and proliferative markers.10

CD4 + T-cells

.—In general, a lower ratio of CD4+ T-cells and T-regs compared with CD8+ T-cells correlates with better prognosis.39 CD4+ T-cells in BrM have been reported to have a hyporesponsive and anergic phenotype.11 Several CD4+ T-cell subclusters have been identified in BrM, namely type 1 T helper-, Th17-, cytotoxic-, IFN-, and T-regs.26 Showing expansion in both tumors and peripheral blood, T-regs have been revealed to induce immunosuppression and coincide with BrM progression.65 Finally, γδ T-cells clustered into 2 main subtypes: IL17-producing and cytotoxic cells,7 and higher γδ T-cells abundance negative correlates with patient survival.39

B-Cells

Evidence regarding the role of BrM-associated B-cells is scarce. In general, infiltration of B-cells is significantly lower than T-cells across cancers of origin.60 High levels of intratumoral plasma cells expressing CD138 have been associated with increased overall survival in patients following resection and radiation.66 In preclinical models, PD1 inhibition increased B-cell proliferation in BrM.67 This is substantiated by evidence indicating a higher abundance of B- and plasma cells in melanoma patients that underwent immunotherapy.57 Moreover, autoantibody levels have been shown to predict BrM.68 Altogether, these changes may suggest a relevant role for B-cell activity during BrM progression and therapy response; however, the functional significance of these changes remains to be elucidated in BrM.

NK Cells and Other Emerging Lymphoid Subsets

NK cells harbor the unique ability to eliminate tumor cells without prior sensitization, positioning them a highly potent component of anticancer immunity. So far, little is known about the role of NK cells in the brain-MME, although animal models have indicated a functional relevance of NK cells because an increase in BrM burden upon NK cell depletion could be observed.69 Latent disseminated cancer cells were shown to hamper NK cell-mediated immunity via downregulation of activators in a WNT-signaling–dependent matter.69

There is growing interest in further lymphoid subsets such as mucosa-associated invariant T (MAIT) cells and innate lymphoid cells (ILCs). Although antigen presentation of microglia to MAIT-cells was documented,70 current evidence on the presence of MAIT-cells in brain neoplasia is largely confined to primary brain tumors.71 There is only limited data on the role of ILCs in BrM, although a study in mice lacking ILC2 showed an increase in CTCs detected in the brain.72 Altogether, while the relevance of these lymphoid subsets in the TME is increasingly recognized, their specific roles in brain-MME remain to be elucidated.

Factors Influencing Immune Variation Illuminated by Differences between Brain-MME and Primary TME

Immune Distinctions between Brain-MME and Primary TME

The brain-MME differs substantially from primary brain-TME such as glioblastoma,11,12 revealing unique BrM-specific features beyond the sole localization in the brain. While primary brain-TME feature high proportions of microglial TAMs, BrM-TAMs largely consist of MDMs.10,53 Remarkably, microglia in BrM can acquire MDM features.11 While notable divergence in microglial phenotypes are observed between primary and metastatic brain tumors, more phenotypic overlap exists in MDMs.11 MDMs adopt a microglia-like phenotype independently of the primary tumor entity,9 likely reflecting adaptation to the localization in the brain. However, monocyte-to-macrophage transition is not homogeneous and is dictated by specific tumor entity.10 Increased vicinity of TAMs to T-cells is also observed in BrM compared with primary brain tumors,11 theoretically allowing for both enhanced antitumor immunity and immunosuppressive activity by TAMs. Neutrophil infiltration and density is elevated in BrM,50,56 as is plasma cell abundance.10 Moreover, T-cell abundance in the brain-MME exceeds that of primary brain-TME, which is limited to sparse perivascular accumulation.69 TCR repertoires in BrM show higher interregional consistency, similar to peripheral TCR repertoires.72 Simultaneously, T-regs accounting for 10% of all CD4+ T-cells constitute a higher proportion of the brain-MME lymphoid compartment compared with brain-TME.10,60 Finally, limited evidence in patient samples suggests that NK cells in the vicinity of BrM form a CD56 expressing, uniquely activated and faster proliferating cluster compared with NK cells in primary brain tumors.10

Besides brain-MME and brain-TME differences, the immune landscape of BrM also contrasts with that of peripheral primary tumors that metastasize to the CNS (peripheral-TME). The immune contrast between brain-MME and peripheral-TME is evident in the lymphoid compartment. CD8+ T-cell numbers are generally diminished in BrM compared with their corresponding primary tumors.44 Notably, a reduced diversity of T-cell clones has been reported in BrM compared with primary lung cancer despite the higher mutational burden found in metastases.73 An increased proportion of NK cells with diminished CD56 expression has been described in BrM compared with primary NSCLC, suggesting an impaired NK population in the metastatic niche.74 Striking differences in B-cell abundance among different cancer entities have been described. Unlike the comparable B-cell compartment in primary and metastatic lung adenocarcinoma, metastases originating from breast cancer showed a stark reduction compared with primary tumors.75 Great variation in the myeloid compartment such as elevated neutrophil infiltration and density in BrM has also been reported.59,60 Comparing TAM subsets in brain-MME and peripheral-TME to elucidate further organ-specific immune interactions will offer insights for new therapeutic strategies. While important, a detailed analysis of MDMs and the resident macrophages (eg, microglia, alveolar, mammary, and dermal) is beyond the scope of this brain-focused review. Understanding the immune landscapes across TMEs as well as various MMEs holds promise for clarifying immune dynamics in BrM and guiding treatments.

Determinants and Modulators of the Immune Composition and Phenotype in BrM

The pronounced differences between brain-MME and primary TME highlight that the brain niche mainly acts as a modulator of the MME, while metastatic tumor cells primarily dictate the immune interactions. Single-cell analyses across patients and primary tumor types in BrM have revealed consistent stromal composition but significant variation in the immune compartment,26 further indicating that metastatic cells from different primary tumor entity are key determinants of the brain-MME composition and phenotype. Immune cell frequency varies across a spectrum encompassing low infiltration levels in breast cancer, higher in lung cancer and strongest in melanoma BrM.10 Concomitantly, BrM stemming from breast cancer were shown to have a higher presence of TAMs, while higher infiltration of lymphocytes was shown in lung cancer (T-cells) and melanoma (B-cells).26 Correlating with a better prognosis,39 increased CD8+ T-cell abundance was most commonly found in melanoma BrM.12 Beyond abundance, infiltration patterns also differ between primary tumor types. Stronger T-cell infiltration at tumor margins than cores has been reported in melanoma,62 while others report predominant stromal infiltration in NSCLC BrM.68 Immune variance in the brain-MME can be determined by specific mutational profiles of metastatic cells. In lung cancer BrM, TP53 mutation has been shown to be associated with a more proliferative transcriptional signature of immune cells and a more immunosuppressive myeloid compartment, particularly TAMs.76 Hypermutated breast cancer BrM, on the other hand, were characterized by a highly inflammatory brain-MME enriched with TAMs, neutrophils, and CD8+ T-cells.76

In addition to the described effects of brain localization and primary tumor entity, further interpatient variance has been described, reporting preferential accumulation of TAMs in the peritumoral region,77 in the tumor mass,21 or both.40 CD8+ T-cells infiltration can range from heavy infiltration to almost complete absence.20 The composition of the immune brain-MME has also been related to features such as patient age, with increased neutrophil, DC, and CD4+ T-cell abundance having been reported in younger patients.12 Finally, previous treatment is believed to contribute to the observed interpatient variance.57,60 Therapeutic interventions such as radiation in late disease stage can transiently shrink the myeloid compartment and shift the TAM composition back to a higher fraction of resident microglia due to increased vulnerability of MDMs.18,57

Targeting the Immune Microenvironment in BrM

Given the lack of adequate therapeutic options for BrM and the dire prognosis of most patients, a pressing need for new therapeutic options exists. Considering the central role of the immune microenvironment in BrM, brain-MME-targeted therapies appear to be a promising approach, and therapeutic interventions targeting TAMs and T-cells have been tested in preclinical and clinical settings.

Targeting TAMs

The dual role of TAMs as both mediators of protumorigenic effects and potential conveyors of antitumor immunity renders them promising therapeutic targets. This is particularly true considering TAM phenotypic plasticity and their intrametastatic localization in vicinity to T-cells.11 In general, two strategies of targeting TAMs have been tested, TAM depletion and TAM reprogramming. TAM depletion by CSF1R inhibitor has been proposed to effectively prevent BrM establishment in a melanoma mouse model.30 However, TAM depletion by CSF1R did not achieve sustained tumor control due to the compensatory upregulation of CSF2R-mediated TAM activation in a mouse model of breast cancer. Targeting the downstream signaling target of CSF2R, STAT5, concomitantly with CSF1R inhibition leads to continued tumor control.25 CSF1R blockade induced a TAM phenotype with similar expression patterns to neurodegenerative disorders, highlighting the detrimental effects of TAM targeting therapies.25 In animal models of several primary tumors, systemic administration of the TLR9 agonist CPG-C triggered microglia to phagocyte, successfully preventing the establishment of BrM.41 Targeting anti-inflammatory TAMs by conditional knock-out of KLF4, which governs the STAT6 pathway, hampered BrM growth in a breast cancer mouse model, similar to pharmacological STAT6 inhibition.78 Activation of STING in TAMs, unlike promising evidence indicating anticancer capacity in models of glioblastoma,79 has not been shown to effectively counter BrM.80 Conversely, STING activation has even been linked to increased occurrence of BrM.81

Finally, in vivo inhibition of the macrophage migration inhibitory factor (MIF), a crucial chemokine for tissue recruitment of TAMs, could synergize with radiotherapy against NSCLC BrM. Blocking MIF signaling via CD74 inhibition could reverse the effect of NSCLC-derived MIF, prompting microglia towards a more migratory, phagocytic phenotype in vitro.82

T-Cell–Based Immunotherapy

Over recent years, classifications characterizing the immunological phenotype of tumors to predict response to immunotherapy have emerged, categorizing tumors on a spectrum from immunologically “hot” to “cold.”83 Primary brain tumors with limited immune infiltration, such as glioblastoma, are defined as “cold” tumors. In contrast, BrM could be quantified as rather “hot” tumors due to the extensive presence of infiltrating lymphocytes and preexisting antitumor immunity, indicated by PD-L1 expression. Consequently, while “cold” tumors such as glioblastoma may benefit from the local introduction of tumor-targeting CAR-T-cells,84 there is a strong rationale for harnessing T-cell immunity with immunotherapies in BrM.

Owing to the high expression of PD1 and PD-L1 in melanoma and breast cancer BrM,26 clinical trials have recently focused on exploring the efficacy of immune checkpoint blockade in BrM, including CTLA4 and PD1 inhibition and their combinations.

A recent phase 2 study reported the investigation of the PD1 antibody pembrolizumab in BrM from a diverse set of untreated and pretreated primary tumors including breast cancer, lung cancer, melanoma, and sarcoma. Intracranial response was observed in around 40% of patients, with selected patients demonstrating survival exceeding 2 years. However, adverse effects were observed in more than 50% of patients.85 Besides, intracranial response to the combination of PD1 inhibitor nivolumab and CTLA4 inhibitor ipilimumab in melanoma BrM was shown to range from 46% to 57% depending on the clinical study but was associated with adverse events in more than half of all patients.86,87 A recent trial of nivolumab and ipilimumab in NSCLC revealed increased 5-year as well as intracranial progression-free survival compared with chemotherapy.88 Encouragingly, a phases II–III trial combining the LAG3-targeting antibody relatlimab and nivolumab demonstrated increased time to melanoma BrM development compared with nivolumab only in a phases II–III trial.89 Preclinical evidence further suggests a synergistic effect of targeted therapies with checkpoint inhibition, combination of the CDK4/6 inhibitor abemaciclib sensitizing melanoma BrM animal models to PD1 inhibition.67

Besides tumor-intrinsic and microenvironmental features, differences in immunotherapy response between BrM and primary brain tumors might also be influenced by the presence of a primary tumor. In a preclinical model of melanoma BrM, the presence of a peripheral tumor was required for intracranial response by increasing CD8+ T-cell trafficking to the BrM.90 There is a possibility that neoantigens expressed by the primary tumor cells may have primed T-cells in the periphery prior to BrM development, potentially enabling these primed T-cells to target the BrM expressing the same neo-antigen.90 Immunotherapy seems to benefit from neo-antigen abundance at the time of treatment, as suggested by improved outcomes of neo-adjuvant over adjuvant immune checkpoint blockade in glioblastoma.91 However, low-reactive T-cells have showed greater overlap with peripheral populations than tumor-specific CD8+ T-cells.60 Therefore, the presence of a primary tumor may influence T-cells beyond direct antigenic priming, supporting the rationale for neo-adjuvant timing of immunotherapy.

The Potential of Combinatory Regimens

Given the significant interindividual variance in the brain-MME immunophenotype, some strongly T-cell infiltrated BrM clearly present as “hot” tumors indicative of a potential response to immunotherapy, other BrM can display the sign of immune exclusion and overall immunosuppressed brain-MME. This variability underscores the rationale for combinatory therapies modulating the brain-MME in addition to strengthening T-cell response.

Existing well-established therapies such as chemoradiation already target cells in the brain-MME beyond tumor cell-intrinsic effects. For instance, radiation has been shown to reduce immunosuppression and increase cytotoxic T-cell numbers.46 Interestingly, the transient change in immune microenvironmental phenotype was subsequently reported to be countered by the increase of immunosuppressive PD-L1 + myeloid cells.46

Chemotherapy in combination with PD-L1 antibody atezolizumab showed intracranial response rates of 45% in NSCLC BrM,92 while combinatory treatment consisting of pembrolizumab and chemotherapy of NSCLC patients with BrM revealed a survival benefit of 12 months compared with chemotherapy alone.93

Combining stereotactic radiosurgery with ipilimumab has further been reported to increase survival in melanoma BrM.94 In line, a recent meta-analysis of both whole brain radiation as well as stereotactic radiosurgery in combination with immunotherapy in melanoma BrM revealed increased survival compared with radiation alone.95 This reported synergy between radiation and immunotherapy likely relates to anticancer effects in nonirradiated brain regions, termed the abscopal effect.96 Underlying mechanisms include radiation-triggered cytokine release and elevated tumor antigen presentation that activates microglia and other immune cells, as well as the elimination of immunosuppressive cells in the microenvironment by radiation.97 Finally, the altered BBB permeability postradiation and subsequent influx of peripheral immune cells as well as efflux of brain-restricted antigens into systemic circulation have been proposed to underlie the abscopal effect.98

While chemotherapy combinations oftentimes aim to achieve highest concurrent doses for maximum anticancer efficacy, the fine-tuned nature of the immune responses elicited by radiation necessitates careful considerations regarding scheduling of radiation and immunotherapy combinatory treatment.97 A trend towards improved prognosis for immunotherapy administered in the first 4 weeks after radiation has been demonstrated.95 Similarly, improved prognosis for patients with melanoma BrM treated with ipilimumab following compared with preceding stereotactic radiosurgery has been shown.99

Combinatory treatment of targeted therapy and immunotherapy in BRAF-mutated melanoma BrM has also been reported to benefit from employing immunotherapy as a follow-up treatment.100 Currently, an on-going clinical trial is investigating combining NK cell therapy and chemotherapy for brain metastatic melanoma (NCT05588453) (Table 1). While early results of TAM and T-cell–directed therapies are promising, investigation of combined approaches that aim at rewiring the brain-MME is still pending. In this regard, novel approaches combining tumor-targeted, pro-tumoral TME-targeted, and immunotherapies could be considered for BrM.

Table 1.

List of prospective clinical trials and retrospective studies investigating combinatorial treatments for brain metastasis.

Identification codePrimary tumor typeTreatment regimenTrial phaseRef.
NCT02374242MelanomaNivolumab + Ipilimumab
vs.
Nivolumab alone
Phase II[86]
NCT02320058MelanomaNivolumab + IpilimumabPhase II[87]
NCT02477826NSCLCFirst-Line Nivolumab + IpilimumabPhase III[88]
NCT03470922MelanomaRelatlimab + Nivolumab
vs.
Nivolumab alone
Phases
II–III
[89]
Atezo-Brain, GECP17/05NSCLCAtezolizumab + Carboplatin + PemetrexedPhase II[92]
NCT02578680NSCLCPemetrexed + Platinum + Pembrolizumab
vs.
Pemetrexed + Platinum + Placebo
Phase III[93]
Retrospective studyMelanomaIpilimumab + Stereotactic Radiosurgery
or
Ipilimumab + Whole Brain Radiotherapy
N/A[94]
Retrospective studyMelanomaStereotactic Radiosurgery + Immunotherapy
or
Whole Brain Radiotherapy + Immunotherapy
vs.
Radiation or Immunotherapy alone
N/A[95]
Retrospective studyMelanomaStereotactic Radiosurgery + IpilimumabN/A[99]
NCT02631447BRAF-mutated metastatic melanomaEncorafenib + Binimetinib
➔ Ipilimumab + Nivolumab
vs.
Ipilimumab + Nivolumab
➔ Encorafenib + Binimetinib
vs.
8-week Encorafenib + Binimetinib
➔ Ipilimumab + Nivolumab
➔ Encorafenib + Binimetinib
Phase II[100]
NCT05588453MelanomaNK Cell Therapy + TemozolomidePhases
I—II
On-going
Identification codePrimary tumor typeTreatment regimenTrial phaseRef.
NCT02374242MelanomaNivolumab + Ipilimumab
vs.
Nivolumab alone
Phase II[86]
NCT02320058MelanomaNivolumab + IpilimumabPhase II[87]
NCT02477826NSCLCFirst-Line Nivolumab + IpilimumabPhase III[88]
NCT03470922MelanomaRelatlimab + Nivolumab
vs.
Nivolumab alone
Phases
II–III
[89]
Atezo-Brain, GECP17/05NSCLCAtezolizumab + Carboplatin + PemetrexedPhase II[92]
NCT02578680NSCLCPemetrexed + Platinum + Pembrolizumab
vs.
Pemetrexed + Platinum + Placebo
Phase III[93]
Retrospective studyMelanomaIpilimumab + Stereotactic Radiosurgery
or
Ipilimumab + Whole Brain Radiotherapy
N/A[94]
Retrospective studyMelanomaStereotactic Radiosurgery + Immunotherapy
or
Whole Brain Radiotherapy + Immunotherapy
vs.
Radiation or Immunotherapy alone
N/A[95]
Retrospective studyMelanomaStereotactic Radiosurgery + IpilimumabN/A[99]
NCT02631447BRAF-mutated metastatic melanomaEncorafenib + Binimetinib
➔ Ipilimumab + Nivolumab
vs.
Ipilimumab + Nivolumab
➔ Encorafenib + Binimetinib
vs.
8-week Encorafenib + Binimetinib
➔ Ipilimumab + Nivolumab
➔ Encorafenib + Binimetinib
Phase II[100]
NCT05588453MelanomaNK Cell Therapy + TemozolomidePhases
I—II
On-going
Table 1.

List of prospective clinical trials and retrospective studies investigating combinatorial treatments for brain metastasis.

Identification codePrimary tumor typeTreatment regimenTrial phaseRef.
NCT02374242MelanomaNivolumab + Ipilimumab
vs.
Nivolumab alone
Phase II[86]
NCT02320058MelanomaNivolumab + IpilimumabPhase II[87]
NCT02477826NSCLCFirst-Line Nivolumab + IpilimumabPhase III[88]
NCT03470922MelanomaRelatlimab + Nivolumab
vs.
Nivolumab alone
Phases
II–III
[89]
Atezo-Brain, GECP17/05NSCLCAtezolizumab + Carboplatin + PemetrexedPhase II[92]
NCT02578680NSCLCPemetrexed + Platinum + Pembrolizumab
vs.
Pemetrexed + Platinum + Placebo
Phase III[93]
Retrospective studyMelanomaIpilimumab + Stereotactic Radiosurgery
or
Ipilimumab + Whole Brain Radiotherapy
N/A[94]
Retrospective studyMelanomaStereotactic Radiosurgery + Immunotherapy
or
Whole Brain Radiotherapy + Immunotherapy
vs.
Radiation or Immunotherapy alone
N/A[95]
Retrospective studyMelanomaStereotactic Radiosurgery + IpilimumabN/A[99]
NCT02631447BRAF-mutated metastatic melanomaEncorafenib + Binimetinib
➔ Ipilimumab + Nivolumab
vs.
Ipilimumab + Nivolumab
➔ Encorafenib + Binimetinib
vs.
8-week Encorafenib + Binimetinib
➔ Ipilimumab + Nivolumab
➔ Encorafenib + Binimetinib
Phase II[100]
NCT05588453MelanomaNK Cell Therapy + TemozolomidePhases
I—II
On-going
Identification codePrimary tumor typeTreatment regimenTrial phaseRef.
NCT02374242MelanomaNivolumab + Ipilimumab
vs.
Nivolumab alone
Phase II[86]
NCT02320058MelanomaNivolumab + IpilimumabPhase II[87]
NCT02477826NSCLCFirst-Line Nivolumab + IpilimumabPhase III[88]
NCT03470922MelanomaRelatlimab + Nivolumab
vs.
Nivolumab alone
Phases
II–III
[89]
Atezo-Brain, GECP17/05NSCLCAtezolizumab + Carboplatin + PemetrexedPhase II[92]
NCT02578680NSCLCPemetrexed + Platinum + Pembrolizumab
vs.
Pemetrexed + Platinum + Placebo
Phase III[93]
Retrospective studyMelanomaIpilimumab + Stereotactic Radiosurgery
or
Ipilimumab + Whole Brain Radiotherapy
N/A[94]
Retrospective studyMelanomaStereotactic Radiosurgery + Immunotherapy
or
Whole Brain Radiotherapy + Immunotherapy
vs.
Radiation or Immunotherapy alone
N/A[95]
Retrospective studyMelanomaStereotactic Radiosurgery + IpilimumabN/A[99]
NCT02631447BRAF-mutated metastatic melanomaEncorafenib + Binimetinib
➔ Ipilimumab + Nivolumab
vs.
Ipilimumab + Nivolumab
➔ Encorafenib + Binimetinib
vs.
8-week Encorafenib + Binimetinib
➔ Ipilimumab + Nivolumab
➔ Encorafenib + Binimetinib
Phase II[100]
NCT05588453MelanomaNK Cell Therapy + TemozolomidePhases
I—II
On-going

Conclusion

A wealth of evidence has indicated the significant role of the immune microenvironmental niche in the pathogenesis of BrM, arising from the complex interactions between tumor cells and surrounding immune cells. While the majority of previous studies considered tumor cells as the central determinants of brain-MME cellular composition and immune condition, the advent of high-throughput single-cell analyses has provided unprecedented insights into the signaling programs within both malignant cells and the bystander immune microenvironment in the metastatic brain tumors. It is increasingly evident that specific tissue conditions can also decisively predefine the susceptibility of a tissue for metastatic invasion as well as contribute significantly to the progression of a tumor. Thus, a thorough understanding of the signaling circuitries within the metastatic immune niche holds great promise to foster the development of novel therapeutic approaches as well as innovative combinatory treatment strategies for patient benefit.

Acknowledgement

All figures were generated with Biorender.com.

Conflict of interest statement. R.B. has received honoraries for lectures and advisory boards from AbbVie, Amgen, AstraZeneca, Bayer, BMS, Boehringer-Ingelheim, Illumina, Janssen, Lilly, Merck-Serono, MSD, Novartis, Qiagen, Pfizer, Roche, Targos MP Inc. R.B. is a co-founder and co-owner of Gnothis Inc./Stockholm (Sweden) and Timer Therapeutics/Freiburg (Germany). All other authors declare no competing interests.

Author contributions

Conceptualization: L.D.S., R.G., P.-H.N. Writing—original draft: L.D.S., P.-H.N. Writing—review and editing: L.D.S., A.F.v.S., S.T.J., M.T., K.-W.N., R.B., H.K., V.N., R.G., P.-H.N. Visualization: A.F.v.S., L.D.S., P.-H.N. Supervision: R.G., P.-H.N.

Funding

P.-H.N., R.B., and H.K. are funded by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) (grant SFB1530-455784452) and by the research network CANcer TARgeting (CANTAR) of the Ministry of Culture and Science of the State of North Rhine-Westphalia (MKW NRW).

References

1.

Berghoff
AS
,
Schur
S
,
Füreder
LM
, et al.
Descriptive statistical analysis of a real life cohort of 2419 patients with brain metastases of solid cancers
.
ESMO Open
.
2016
;
1
(
2
):
e000024
.

2.

Jünger
ST
,
Pennig
L
,
Schödel
P
, et al.
The debatable benefit of gross-total resection of brain metastases in a comprehensive treatment setting
.
Cancers
.
2021
;
13
(
6
):
1435
.

3.

Kocher
M
,
Soffietti
R
,
Abacioglu
U
, et al.
Adjuvant whole-brain radiotherapy versus observation after radiosurgery or surgical resection of one to three cerebral metastases: results of the EORTC 22952-26001 Study
.
J Clin Oncol.
2011
;
29
(
2
):
134
141
.

4.

Le Rhun
E
,
Guckenberger
M
,
Smits
M
, et al.
EANO–ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up of patients with brain metastasis from solid tumours
.
Ann Oncol.
2021
;
32
(
11
):
1332
1347
.

5.

Cagney
DN
,
Martin
AM
,
Catalano
PJ
, et al.
Incidence and prognosis of patients with brain metastases at diagnosis of systemic malignancy: a population-based study
.
Neuro-Oncology.
2017
;
19
(
11
):
1511
1521
.

6.

Quail
DF
,
Joyce
JA.
The Microenvironmental Landscape of Brain Tumors
.
Cancer Cell
.
2017
;
31
(
3
):
326
341
.

7.

Mirzaei
R
,
Yong
VW.
Microglia–T cell conversations in brain cancer progression
.
Trends Mol Med.
2022
;
28
(
11
):
951
963
.

8.

Fischer
GM
,
Jalali
A
,
Kircher
DA
, et al.
Molecular profiling reveals unique immune and metabolic features of melanoma brain metastases
.
Cancer Discov
.
2019
;
9
(
5
):
628
645
.

9.

Andersen
BM
,
Faust Akl
C
,
Wheeler
MA
, et al.
Glial and myeloid heterogeneity in the brain tumour microenvironment
.
Nat Rev Cancer.
2021
;
21
(
12
):
786
802
.

10.

Friebel
E
,
Kapolou
K
,
Unger
S
, et al.
Single-cell mapping of human brain cancer reveals tumor-specific instruction of tissue-invading leukocytes
.
Cell.
2020
;
181
(
7
):
1626
1642.e20
.

11.

Klemm
F
,
Maas
RR
,
Bowman
RL
, et al.
Interrogation of the microenvironmental landscape in brain tumors reveals disease-specific alterations of immune cells
.
Cell.
2020
;
181
(
7
):
1643
1660.e17
.

12.

Karimi
E
,
Yu
MW
,
Maritan
SM
, et al.
Single-cell spatial immune landscapes of primary and metastatic brain tumours
.
Nature.
2023
;
614
(
7948
):
555
563
.

13.

Sankowski
R
,
Böttcher
C
,
Masuda
T
, et al.
Mapping microglia states in the human brain through the integration of high-dimensional techniques
.
Nat Neurosci.
2019
;
22
(
12
):
2098
2110
.

14.

Van Hove
H
,
Martens
L
,
Scheyltjens
I
, et al.
A single-cell atlas of mouse brain macrophages reveals unique transcriptional identities shaped by ontogeny and tissue environment
.
Nat Neurosci.
2019
;
22
(
6
):
1021
1035
.

15.

Chuang
H
,
Van Rossum
D
,
Sieger
D
, et al.
Carcinoma cells misuse the host tissue damage response to invade the brain
.
Glia.
2013
;
61
(
8
):
1331
1346
.

16.

Bowman
RL
,
Klemm
F
,
Akkari
L
, et al.
Macrophage ontogeny underlies differences in tumor-specific education in brain malignancies
.
Cell Rep
.
2016
;
17
(
9
):
2445
2459
.

17.

Guldner
IH
,
Wang
Q
,
Yang
L
, et al.
CNS-native myeloid cells drive immune suppression in the brain metastatic niche through Cxcl10
.
Cell.
2020
;
183
(
5
):
1234
1248.e25
.

18.

Schulz
M
,
Michels
B
,
Niesel
K
, et al.
Cellular and molecular changes of brain metastases-associated myeloid cells during disease progression and therapeutic response
.
iScience
.
2020
;
23
(
6
):
101178
.

19.

Zhang
W
,
Karschnia
P
,
von Mücke-Heim
IA
, et al.
In vivo two-photon characterization of tumor-associated macrophages and microglia (TAM/M) and CX3CR1 during different steps of brain metastasis formation from lung cancer
.
Neoplasia
.
2021
;
23
(
11
):
1089
1100
.

20.

Rubio-Perez
C
,
Planas-Rigol
E
,
Trincado
JL
, et al.
Immune cell profiling of the cerebrospinal fluid enables the characterization of the brain metastasis microenvironment
.
Nat Commun.
2021
;
12
(
1
):
1503
.

21.

Lorger
M
,
Felding-Habermann
B.
Capturing changes in the brain microenvironment during initial steps of breast cancer brain metastasis
.
Am J Pathol.
2010
;
176
(
6
):
2958
2971
.

22.

Zhang
Q
,
Abdo
R
,
Iosef
C
, et al.
The spatial transcriptomic landscape of non-small cell lung cancer brain metastasis
.
Nat Commun.
2022
;
13
(
1
):
5983
.

23.

Evans
KT
,
Blake
K
,
Longworth
A
, et al.
Microglia promote anti-tumour immunity and suppress breast cancer brain metastasis
.
Nat Cell Biol.
2023
;
25
(
12
):
1848
1859
.

24.

Zou
Y
,
Ye
F
,
Kong
Y
, et al.
The single-cell landscape of intratumoral heterogeneity and the immunosuppressive microenvironment in liver and brain metastases of breast cancer
.
Adv Sci.
2023
;
10
(
5
):
2203699
.

25.

Klemm
F
,
Möckl
A
,
Salamero-Boix
A
, et al.
Compensatory CSF2-driven macrophage activation promotes adaptive resistance to CSF1R inhibition in breast-to-brain metastasis
.
Nat Cancer
.
2021
;
2
(
10
):
1086
1101
.

26.

Gonzalez
H
,
Mei
W
,
Robles
I
, et al.
Cellular architecture of human brain metastases
.
Cell.
2022
;
185
(
4
):
729
745.e20
.

27.

Gan
DX
,
Wang
YB
,
He
MY
, et al.
Lung cancer cells-controlled dkk-1 production in brain metastatic cascade drive microglia to acquire a pro-tumorigenic phenotype
.
Front Cell Dev Biol.
2020
;
8
:
591405
.

28.

Foo
SL
,
Sachaphibulkij
K
,
Lee
CLY
, et al.
Breast cancer metastasis to brain results in recruitment and activation of microglia through annexin-A1/formyl peptide receptor signaling
.
Breast Cancer Res.
2022
;
24
(
1
):
25
.

29.

Jin
Y
,
Kang
Y
,
Wang
M
, et al.
Targeting polarized phenotype of microglia via IL6/JAK2/STAT3 signaling to reduce NSCLC brain metastasis
.
Sig Transduct Target Ther
.
2022
;
7
(
1
):
52
.

30.

Qiao
S
,
Qian
Y
,
Xu
G
,
Luo
Q
,
Zhang
Z.
Long-term characterization of activated microglia/macrophages facilitating the development of experimental brain metastasis through intravital microscopic imaging
.
J Neuroinflammation.
2019
;
16
(
1
):
4
.

31.

Andreou
KE
,
Soto
MS
,
Allen
D
, et al.
Anti-inflammatory microglia/macrophages as a potential therapeutic target in brain metastasis
.
Front Oncol.
2017
;
7
:
251
.

32.

Pukrop
T
,
Dehghani
F
,
Chuang
H
, et al.
Microglia promote colonization of brain tissue by breast cancer cells in a Wnt-dependent way
.
Glia.
2010
;
58
(
12
):
1477
1489
.

33.

Berghoff
AS
,
Rajky
O
,
Winkler
F
, et al.
Invasion patterns in brain metastases of solid cancers
.
Neuro-Oncology.
2013
;
15
(
12
):
1664
1672
.

34.

Priego
N
,
Zhu
L
,
Monteiro
C
, et al.
STAT3 labels a subpopulation of reactive astrocytes required for brain metastasis
.
Nat Med.
2018
;
24
(
7
):
1024
1035
.

35.

Blazquez
R
,
Wlochowitz
D
,
Wolff
A
, et al.
PI3K: A master regulator of brain metastasis-promoting macrophages/microglia
.
Glia.
2018
;
66
(
11
):
2438
2455
.

36.

Sevenich
L
,
Bowman
RL
,
Mason
SD
, et al.
Analysis of tumour- and stroma-supplied proteolytic networks reveals a brain-metastasis-promoting role for cathepsin S
.
Nat Cell Biol.
2014
;
16
(
9
):
876
888
.

37.

Zhang
L
,
Zhang
S
,
Yao
J
, et al.
Microenvironment-induced PTEN loss by exosomal microRNA primes brain metastasis outgrowth
.
Nature.
2015
;
527
(
7576
):
100
104
.

38.

Fitzgerald
DP
,
Palmieri
D
,
Hua
E
, et al.
Reactive glia are recruited by highly proliferative brain metastases of breast cancer and promote tumor cell colonization
.
Clin Exp Metastasis.
2008
;
25
(
7
):
799
810
.

39.

Griguolo
G
,
Tosi
A
,
Dieci
MV
, et al.
A comprehensive profiling of the immune microenvironment of breast cancer brain metastases
.
Neuro-Oncology.
2022
;
24
(
12
):
2146
2158
.

40.

He
BP
,
Wang
JJ
,
Zhang
X
, et al.
Differential reactions of microglia to brain metastasis of lung cancer
.
Mol Med.
2006
;
12
(
7-8
):
161
170
.

41.

Benbenishty
A
,
Gadrich
M
,
Cottarelli
A
, et al.
Prophylactic TLR9 stimulation reduces brain metastasis through microglia activation
.
PLoS Biol
.
2019
;
17
(
3
):
e2006859
.

42.

Wu
SY
,
Sharma
S
,
Wu
K
, et al.
Tamoxifen suppresses brain metastasis of estrogen receptor-deficient breast cancer by skewing microglia polarization and enhancing their immune functions
.
Breast Cancer Res.
2021
;
23
(
1
):
35
.

43.

Moshe
A
,
Izraely
S
,
Sagi-Assif
O
, et al.
Cystatin C takes part in melanoma-microglia cross-talk: possible implications for brain metastasis
.
Clin Exp Metastasis.
2018
;
35
(
5-6
):
369
378
.

44.

Herrera-Rios
D
,
Mughal
SS
,
Teuber-Hanselmann
S
, et al.
Macrophages/microglia represent the major source of indolamine 2,3-dioxygenase expression in melanoma metastases of the brain
.
Front Immunol.
2020
;
11
:
120
.

45.

Wainwright
DA
,
Balyasnikova
IV
,
Chang
AL
, et al.
IDO expression in brain tumors increases the recruitment of regulatory T cells and negatively impacts survival
.
Clin Cancer Res.
2012
;
18
(
22
):
6110
6121
.

46.

Niesel
K
,
Schulz
M
,
Anthes
J
, et al.
The immune suppressive microenvironment affects efficacy of radio-immunotherapy in brain metastasis
.
EMBO Mol Med.
2021
;
13
(
5
):
e13412
.

47.

Louie
E
,
Chen
XF
,
Coomes
A
, et al.
Neurotrophin-3 modulates breast cancer cells and the microenvironment to promote the growth of breast cancer brain metastasis
.
Oncogene.
2013
;
32
(
35
):
4064
4077
.

48.

Kleffman
K
,
Levinson
G
,
Rose
IVL
, et al.
Melanoma-secreted amyloid beta suppresses neuroinflammation and promotes brain metastasis
.
Cancer Discov
.
2022
;
12
(
5
):
1314
1335
.

49.

Daoud
M
,
Broxtermann
PN
,
Schorn
F
, et al.
XIAP promotes melanoma growth by inducing tumour neutrophil infiltration
.
EMBO Rep.
2022
;
23
(
6
):
e53608
.

50.

Maas
RR
,
Soukup
K
,
Fournier
N
, et al.
The local microenvironment drives activation of neutrophils in human brain tumors
.
Cell.
2023
;
186
(
21
):
4546
4566.e27
.

51.

Mitsuya
K
,
Nakasu
Y
,
Kurakane
T
, et al.
Elevated preoperative neutrophil-to-lymphocyte ratio as a predictor of worse survival after resection in patients with brain metastasis
.
J Neurosurg.
2017
;
127
(
2
):
433
437
.

52.

Zhang
L
,
Yao
J
,
Wei
Y
, et al.
Blocking immunosuppressive neutrophils deters pY696-EZH2–driven brain metastases
.
Sci Transl Med.
2020
;
12
(
545
):
eaaz5387
.

53.

Sun
H
,
Li
L
,
Lao
I
, et al.
Single-cell RNA sequencing reveals cellular and molecular reprograming landscape of gliomas and lung cancer brain metastases
.
Clin Transl Med
.
2022
;
12
(
11
):
e1101
.

54.

Adler
O
,
Zait
Y
,
Cohen
N
, et al.
Reciprocal interactions between innate immune cells and astrocytes facilitate neuroinflammation and brain metastasis via lipocalin-2
.
Nat Cancer
.
2023
;
4
(
3
):
401
418
.

55.

Liu
Y
,
Kosaka
A
,
Ikeura
M
, et al.
Premetastatic soil and prevention of breast cancer brain metastasis
.
Neuro-Oncology.
2013
;
15
(
7
):
891
903
.

56.

Souza
VGP
,
Forder
A
,
Telkar
N
, et al.
Identifying new contributors to brain metastasis in lung adenocarcinoma: a transcriptomic meta-analysis
.
Cancers
.
2023
;
15
(
18
):
4526
.

57.

Smalley
I
,
Chen
Z
,
Phadke
M
, et al.
Single-cell characterization of the immune microenvironment of melanoma brain and leptomeningeal metastases
.
Clin Cancer Res.
2021
;
27
(
14
):
4109
4125
.

58.

Korin
B
,
Ben-Shaanan
TL
,
Schiller
M
, et al.
High-dimensional, single-cell characterization of the brain’s immune compartment
.
Nat Neurosci.
2017
;
20
(
9
):
1300
1309
.

59.

Fukumura
K
,
Malgulwar
PB
,
Fischer
GM
, et al.
Multi-omic molecular profiling reveals potentially targetable abnormalities shared across multiple histologies of brain metastasis
.
Acta Neuropathol.
2021
;
141
(
2
):
303
321
.

60.

Sudmeier
LJ
,
Hoang
KB
,
Nduom
EK
, et al.
Distinct phenotypic states and spatial distribution of CD8+ T cell clonotypes in human brain metastases
.
Cell Rep Med
.
2022
;
3
(
5
):
100620
.

61.

Soto
MS
,
Serres
S
,
Anthony
DC
,
Sibson
NR.
Functional role of endothelial adhesion molecules in the early stages of brain metastasis
.
Neuro-Oncology.
2014
;
16
(
4
):
540
551
.

62.

Amit
M
,
Laider-Trejo
L
,
Shalom
V
, et al.
Characterization of the melanoma brain metastatic niche in mice and humans
.
Cancer Med
.
2013
;
2
(
2
):
155
163
.

63.

Wischnewski
V
,
Maas
RR
,
Aruffo
PG
, et al.
Phenotypic diversity of T cells in human primary and metastatic brain tumors revealed by multiomic interrogation
.
Nat Cancer
.
2023
;
4
(
6
):
908
924
.

64.

Biermann
J
,
Melms
JC
,
Amin
AD
, et al.
Dissecting the treatment-naive ecosystem of human melanoma brain metastasis
.
Cell.
2022
;
185
(
14
):
2591
2608.e30
.

65.

Jacobs
JFM
,
Idema
AJ
,
Bol
KF
, et al.
Regulatory T cells and the PD-L1/PD-1 pathway mediate immune suppression in malignant human brain tumors
.
Neuro-Oncology.
2009
;
11
(
4
):
394
402
.

66.

Soike
MH
,
Logue
J
,
Qasem
S
, et al.
CD138 plasma cells may predict brain metastasis recurrence following resection and stereotactic radiosurgery
.
Sci Rep.
2019
;
9
(
1
):
14385
.

67.

Nayyar
N
,
De Sauvage
MA
,
Chuprin
J
, et al.
CDK4/6 inhibition sensitizes intracranial tumors to PD-1 blockade in preclinical models of brain metastasis
.
Clin Cancer Res.
2024
;
30
(
2
):
420
435
.

68.

Choi
H
,
Puvenna
V
,
Brennan
C
, et al.
S100B and S100B autoantibody as biomarkers for early detection of brain metastases in lung cancer
.
Transl Lung Cancer Res
.
2016
;
5
(
4
):
413
419
.

69.

Malladi
S
,
Macalinao
DG
,
Jin
X
, et al.
Metastatic latency and immune evasion through autocrine inhibition of WNT
.
Cell.
2016
;
165
(
1
):
45
60
.

70.

Priya
R
,
Brutkiewicz
RR.
Brain astrocytes and microglia express functional MR1 molecules that present microbial antigens to mucosal-associated invariant T (MAIT) cells
.
J Neuroimmunol.
2020
;
349
:
577428
.

71.

Peterfalvi
A
,
Gomori
E
,
Magyarlaki
T
, et al.
Invariant V 7.2-J 33 TCR is expressed in human kidney and brain tumors indicating infiltration by mucosal-associated invariant T (MAIT) cells
.
Int Immunol.
2008
;
20
(
12
):
1517
1525
.

72.

Saranchova
I
,
Han
J
,
Zaman
R
, et al.
Type 2 innate lymphocytes actuate immunity against tumours and limit cancer metastasis
.
Sci Rep.
2018
;
8
(
1
):
2924
.

73.

Mansfield
AS
,
Ren
H
,
Sutor
S
, et al.
Contraction of T cell richness in lung cancer brain metastases
.
Sci Rep.
2018
;
8
(
1
):
2171
.

74.

Song
SG
,
Kim
S
,
Koh
J
, et al.
Comparative analysis of the tumor immune-microenvironment of primary and brain metastases of non-small-cell lung cancer reveals organ-specific and EGFR mutation-dependent unique immune landscape
.
Cancer Immunol Immunother.
2021
;
70
(
7
):
2035
2048
.

75.

Cimino-Mathews
A
,
Ye
X
,
Meeker
A
,
Argani
P
,
Emens
LA.
Metastatic triple-negative breast cancers at first relapse have fewer tumor-infiltrating lymphocytes than their matched primary breast tumors: a pilot study
.
Hum Pathol.
2013
;
44
(
10
):
2055
2063
.

76.

Álvarez-Prado
AF
,
Maas
RR
,
Soukup
K
, et al.
Immunogenomic analysis of human brain metastases reveals diverse immune landscapes across genetically distinct tumors
.
Cell Rep Med
.
2023
;
4
(
1
):
100900
.

77.

Zhang
M
,
Olsson
Y.
Reactions of astrocytes and microglial cells around hematogenous metastases of the human brain Expression of endothelin-like immunoreactivity in reactive astrocytes and activation of microglial cells
.
J Neurol Sci.
1995
;
134
(
1-2
):
26
32
.

78.

Economopoulos
V
,
Pannell
M
,
Johanssen
VA
, et al.
Inhibition of anti-inflammatory macrophage phenotype reduces tumour growth in mouse models of brain metastasis
.
Front Oncol.
2022
;
12
:
850656
.

79.

Berger
G
,
Knelson
EH
,
Jimenez-Macias
JL
, et al.
STING activation promotes robust immune response and NK cell–mediated tumor regression in glioblastoma models
.
Proc Natl Acad Sci USA.
2022
;
119
(
28
):
e2111003119
.

80.

Bähr
O
,
Gross
S
,
Harter
P
, et al.
ASA404, a vascular disrupting agent, as an experimental treatment approach for brain tumors
.
Oncol Lett
.
2017
;
14
(
5
):
5443
5451
.

81.

Low
JT
,
Brown
MC
,
Reitman
ZJ
, et al.
Understanding and therapeutically exploiting cGAS/STING signaling in glioblastoma
.
J Clin Investig.
2024
;
134
(
2
):
e163452
.

82.

Liu
L
,
Wang
J
,
Wang
Y
, et al.
Blocking the MIF-CD74 axis augments radiotherapy efficacy for brain metastasis in NSCLC via synergistically promoting microglia M1 polarization
.
J Exp Clin Cancer Res.
2024
;
43
(
1
):
128
.

83.

Galon
J
,
Bruni
D.
Approaches to treat immune hot, altered and cold tumours with combination immunotherapies
.
Nat Rev Drug Discov.
2019
;
18
(
3
):
197
218
.

84.

Choi
BD
,
Gerstner
ER
,
Frigault
MJ
, et al.
Intraventricular CARv3-TEAM-E T Cells in Recurrent Glioblastoma
.
N Engl J Med.
2024
;
390
(
14
):
1290
1298
.

85.

Brastianos
PK
,
Kim
AE
,
Giobbie-Hurder
A
, et al.
Pembrolizumab in brain metastases of diverse histologies: phase 2 trial results
.
Nat Med.
2023
;
29
(
7
):
1728
1737
.

86.

Long
GV
,
Atkinson
V
,
Lo
S
, et al.
Combination nivolumab and ipilimumab or nivolumab alone in melanoma brain metastases: a multicentre randomised phase 2 study
.
Lancet Oncol.
2018
;
19
(
5
):
672
681
.

87.

Tawbi
HA
,
Forsyth
PA
,
Algazi
A
, et al.
Combined Nivolumab and Ipilimumab in Melanoma Metastatic to the Brain
.
N Engl J Med.
2018
;
379
(
8
):
722
730
.

88.

Reck
M
,
Ciuleanu
TE
,
Lee
JS
, et al.
Systemic and intracranial outcomes with first-line nivolumab plus ipilimumab in patients with metastatic NSCLC and baseline brain metastases from CheckMate 227 Part 1
.
J Thorac Oncol.
2023
;
18
(
8
):
1055
1069
.

89.

Tawbi
HA
,
Schadendorf
D
,
Lipson
EJ
, et al.
Relatlimab and Nivolumab versus Nivolumab in Untreated Advanced Melanoma
.
N Engl J Med.
2022
;
386
(
1
):
24
34
.

90.

Taggart
D
,
Andreou
T
,
Scott
KJ
, et al.
Anti–PD-1/anti–CTLA-4 efficacy in melanoma brain metastases depends on extracranial disease and augmentation of CD8 + T cell trafficking
.
Proc Natl Acad Sci USA.
2018
;
115
(
7
):
E1540
E1549
.

91.

Cloughesy
TF
,
Mochizuki
AY
,
Orpilla
JR
, et al.
Neoadjuvant anti-PD-1 immunotherapy promotes a survival benefit with intratumoral and systemic immune responses in recurrent glioblastoma
.
Nat Med.
2019
;
25
(
3
):
477
486
.

92.

Nadal
E
,
Rodríguez-Abreu
D
,
Simó
M
, et al.
Phase II trial of atezolizumab combined with carboplatin and pemetrexed for patients with advanced nonsquamous non–small-cell lung cancer with untreated brain metastases (Atezo-Brain, GECP17/05)
.
JCO.
2023
;
41
(
28
):
4478
4485
.

93.

Gadgeel
S
,
Rodríguez-Abreu
D
,
Speranza
G
, et al.
Updated Analysis From KEYNOTE-189: Pembrolizumab or placebo plus pemetrexed and platinum for previously untreated metastatic nonsquamous non–small-cell lung cancer
.
JCO.
2020
;
38
(
14
):
1505
1517
.

94.

Silk
AW
,
Bassetti
MF
,
West
BT
,
Tsien
CI
,
Lao
CD.
Ipilimumab and radiation therapy for melanoma brain metastases
.
Cancer Med
.
2013
;
2
(
6
):
899
906
.

95.

Moyers
JT
,
Chong
EG
,
Peng
J
, et al.
Real world outcomes of combination and timing of immunotherapy with radiotherapy for melanoma with brain metastases
.
Cancer Med
.
2021
;
10
(
4
):
1201
1211
.

96.

Tracz
JA
,
Donnelly
BM
,
Ngu
S
, et al.
The abscopal effect: inducing immunogenicity in the treatment of brain metastases secondary to lung cancer and melanoma
.
J Neurooncol.
2023
;
163
(
1
):
1
14
.

97.

Galluzzi
L
,
Aryankalayil
MJ
,
Coleman
CN
,
Formenti
SC.
Emerging evidence for adapting radiotherapy to immunotherapy
.
Nat Rev Clin Oncol.
2023
;
20
(
8
):
543
557
.

98.

Pangal
DJ
,
Yarovinsky
B
,
Cardinal
T
, et al.
The abscopal effect: systematic review in patients with brain and spine metastases
.
Neurooncol Adv.
2022
;
4
(
1
):
vdac132
.

99.

Kiess
AP
,
Wolchok
JD
,
Barker
CA
, et al.
Stereotactic radiosurgery for melanoma brain metastases in patients receiving ipilimumab: safety profile and efficacy of combined treatment
.
Int J Radiat Oncol Biol Phys.
2015
;
92
(
2
):
368
375
.

100.

Ascierto
PA
,
Mandalà
M
,
Ferrucci
PF
, et al.
Sequencing of ipilimumab plus nivolumab and encorafenib plus binimetinib for untreated BRAF-mutated metastatic melanoma (SECOMBIT): a randomized, three-arm, open-label phase II trial
.
JCO.
2023
;
41
(
2
):
212
221
.

This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial License (https://creativecommons.org/licenses/by-nc/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited. For commercial re-use, please contact [email protected] for reprints and translation rights for reprints. All other permissions can be obtained through our RightsLink service via the Permissions link on the article page on our site—for further information please contact [email protected].